EP0612250A1 - PROCEDES ET COMPOSITIONS RELATIFS A DES ANTIGENES UTILES DE -i(MORAXELLA CATARRHALIS). - Google Patents

PROCEDES ET COMPOSITIONS RELATIFS A DES ANTIGENES UTILES DE -i(MORAXELLA CATARRHALIS).

Info

Publication number
EP0612250A1
EP0612250A1 EP92918273A EP92918273A EP0612250A1 EP 0612250 A1 EP0612250 A1 EP 0612250A1 EP 92918273 A EP92918273 A EP 92918273A EP 92918273 A EP92918273 A EP 92918273A EP 0612250 A1 EP0612250 A1 EP 0612250A1
Authority
EP
European Patent Office
Prior art keywords
antigen
antibody
omp
catarrhalis
accordance
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
EP92918273A
Other languages
German (de)
English (en)
Other versions
EP0612250B1 (fr
Inventor
Eric J Hansen
Merja Helminen
Isobel Maciver
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of Texas System
Original Assignee
University of Texas System
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University of Texas System filed Critical University of Texas System
Publication of EP0612250A1 publication Critical patent/EP0612250A1/fr
Application granted granted Critical
Publication of EP0612250B1 publication Critical patent/EP0612250B1/fr
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/12Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria
    • C07K16/1203Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-negative bacteria
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/21Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Pseudomonadaceae (F)
    • C07K14/212Moraxellaceae, e.g. Acinetobacter, Moraxella, Oligella, Psychrobacter
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/12Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria
    • C07K16/1203Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-negative bacteria
    • C07K16/1217Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-negative bacteria from Neisseriaceae (F)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies

Definitions

  • the present invention relates generally to various outer membrane proteins (OMPs) of Moraxella catarrhalis which have been found by the inventors to be useful targets in immunotherapy, such as in the preparation of vaccines or protective antibodies for use in treatment of Moraxella catarrhalis-related diseases.
  • OMPs outer membrane proteins
  • the present invention concerns antigens identified by molecular weights of about 30 kD, 80 kD and a third antigen, termed "high molecular weight protein" or "HMWP" antigen having a molecular weight of between about 200 and 700 kD, as measured by SDS-polya ⁇ rylamide gel electrophoresis.
  • the invention concerns recombinant clones encoding these antigens, antigen fragments derived therefrom, equivalents thereof, as well as to antibodies reactive with these species. Further, the invention concerns methods for the detection of Moraxella catarrhalis antigens and antibodies, as well as the use of specific antigens and antibodies both in passive and active immunity against Moraxella catarrhalis infections.
  • Moraxella catarrhalis (previously known as Branhamella catarrhalis or Neisseria catarrhalis) was a harmless saprophyte of the upper respiratory tract.
  • this organism is an important hu an pathogen.
  • Moraxella catarrhalis is a leading cause of otitis media, acute maxillary sinusitis as well as generalized infections of the lower respiratory tract (see, e.g.. Murphy et al . , 1989).
  • Moraxella catarrhalis antigens that would serve as potentially important targets of the human immune response to infection (Murphy, 1989; Goldblatt et al . , 1990; Murphy et al . , 1990).
  • OMPs outer membrane proteins
  • LOS lipooligo-saccharide
  • fimbriae the surface of Moraxella catarrhalis is composed of outer membrane proteins (OMPs) , lipooligo-saccharide (LOS) and fimbriae.
  • OMPs outer membrane proteins
  • LOS lipooligo-saccharide
  • LOS The LOS of Moraxella catarrhalis has also been suggested as a possible target for vaccine development. LOS has been isolated from Moraxella catarrhalis strains and subjected to SDS-PAGE and silver staining (Murphy,
  • Moraxella catarrhalis component may serve as useful antigens that can, for example, be employed in the preparation of both passive and active immunotherapeutic reagents such as vaccines. Additionally, once such an antigen or antigens is identified, there is a need for providing methods and compositions which will allow the preparation of these vaccines and quantities that will allow their use on a wide scale basis in therapeutic protocols.
  • the present invention is concerned with the identification and subsequent preparation of an Moraxella catarrhalis antigen species that would be of use both in the prevention and diagnosis of disease.
  • the invention concerns the inventors' surprising discovery that particular Moraxella catarrhalis OMP antigens, including the 30 kD, 80 kD and HMWP OMP antigens, have particular utility in vaccine development. It is postulated by the inventors, therefore, these antigens can be used directly as a component of a vaccine, or can be employed for the preparation of corresponding or equivalent antigen through sequence analysis.
  • the present invention thus concerns an antigen composition
  • an antigen composition comprising a purified protein or peptide antigen incorporating an epitope that is immunologically cross-reactive with one or more of the foregoing M. catarrhalis OMP antigens.
  • the purified protein or peptide antigen will comprise the OMP itself
  • the present disclosure provides techniques which may be employed for preparing variants of these OMP antigens, peptides that incorporate related antigenic epitopes, as well as antigenic functional equivalents of each of these.
  • DNA segments encoding the various OMP antigens are disclosed, the antigens may be provided essentially free of antigenic epitopes from other M.
  • catarrhalis antigens through the application of recombinant technology. That is, one may prepare the antigen by recombinant expression means using a host cell other than M. catarrhalis or related species, and thereby provide the antigen in an essentially pure antigenic state, with respect to other M. catarrhalis antigens. Such preparations will therefore be free, e . g. , of LOS or fimbriae antigens.
  • DNA segments disclosed herein may be sequenced, and from this DNA sequence one may determine the underlying amino acid sequence of the selected OMP protein, whether it be the 30 kD, 80 kD or HMWP OMP species. Once this information is obtained, identification of suitable antigenic epitopes is a relatively straightforward matter through the use of, for example, software programs for the prediction of such epitopes that are available to those of skill in the art. The amino acid sequence of these "epitopic core sequences" may then be readily incorporated into shorter peptides, either through the application of peptide synthesis or recombinant technology.
  • Preferred peptides will generally be on the order of 15 to 50 amino acids in length, and more preferably about 15 to about 30 amino acids in length. It is proposed that shorter antigenic peptides which incorporate epitopes of the selected OMP will provide advantages in certain circumstances, for example, in the preparation of vaccines or in immunologic detection assays. Exemplary advantages include the ability to circumvent problems of contamination and purity often associated with proteins prepared by recombinant production in that peptides of this length may be prepared readily be synthetic means using peptide synthesizers.
  • the present invention concerns processes for preparing compositions which include purified protein or peptide antigens that incorporate epitopes that are immunologically cross-reactive with the 30 kD, 80 kD or HMWP OMP.
  • these processes include first selecting cells that are capable of expressing such a protein or peptide antigen, culturing the cells under conditions effective to allow expression of the antigen, and collecting the antigen to thereby prepare the composition.
  • one desires to prepare the OMP antigen itself one will simply desire to culture M. catarrhalis cells as a first step. In this case, the antigen will be provided, upon expression, in the outer membrane fraction of the cell.
  • the antigen is then prepared by, first, preparation of membrane fraction followed by solubilization and extraction of the antigen from the prepared membranes using an ionic or non-ionic detergent. Further purification may be achieved by a variety of methods including column fractionation, isoelectric focusing, and the like, or even immunoadsorption employing OMP-directed antibodies.
  • Preferred recombinant host cells for expression of antigens in accordance with the invention will typically be a bacterial host cell in that the antigen is a bacterial antigen.
  • Preferred bacterial host cells include E. coli , H. influenzae , Salmonella species, Mycobacterium species, or even Bacillis subtilis cells.
  • the present invention concerns DNA segments which encode the desire protein or peptide antigen. Methods are disclosed herein for obtaining such segments in a purified state relative to their naturally occurring state. These DNA segments will have a number of advantages and uses. For exa ple, segments encoding the entire OMP gene may be introduced into recombinant host cells and employed for expressing the entire protein antigen. Alternatively, through the application of genetic engineering techniques, subportions or derivatives of the selected OMP gene may be employed to prepare shorter peptide sequences which nevertheless incorporate the desired antigenic epitopes. Furthermore, through the application of site-directed mutagenesis techniques, one may re- engineer DNA segments of the present invention to alter the coding sequence, e .g .
  • antigenically functional equivalent peptides to introduce improvements to the antigenicity of epitopic core sequences and thereby prepare antigenically functional equivalent peptides.
  • antigenically functional equivalent peptides may also prepare fusion peptides, e.g., where the antigen coding regions are aligned within the same expression unit with other desired antigen or proteins or peptides having desired functions, such as for immunodetection purposes (e.g., enzyme label coding regions) .
  • DNA segments of the present invention are incorporated into appropriate vector sequences which may, e.g., improve the efficiency of transfection of host cells.
  • bacterial host cells it is proposed that virtually any vector known in the art to be appropriate for the selected host cell may be employed.
  • plasmid vectors such as pBR322, or bacteriophages such as ⁇ GEM-
  • nucleic acid segments of the present invention will have numerous uses other than in connection with expression of antigenic peptides or proteins.
  • nucleic acid segments of at least 14 or so nucleotides in length that incorporate regions of the OMP gene sequence may be employed as selective hybridization probes for the detection of M. catarrhalis sequences in selected samples or, e.g., to screen clone banks to identify clones which comprise corresponding or related sequences.
  • short segments may be employed as nucleic acid primers, such as in connection with PCR technology, for use in any of a number of applications, including, e.g., cloning and engineering exercises, or in PCR-based detection protocols.
  • the invention concerns the preparation of antibodies capable of immunocomplexing with epitopes of the OMP antigen.
  • Particular techniques for preparing antibodies in accordance with the invention are disclosed hereinbelow.
  • any of the current techniques known in the art for the preparation of antibodies in general may be employed, through the application of either monoclonal or polyclonal technology.
  • a surprising aspect of the invention involves the inventors' discovery that monoclonal antibodies directed against the 30 kD, 80 kD and HMWP OMP antigens provide a protective effect against M . catarrhalis challenge in animal models.
  • the present invention is directed both to vaccine compositions which include an antigen in accordance with the present invention, or antibodies against such an antigen, together with a pharmaceutically acceptable carrier, diluent, or adjuvant.
  • the present invention concerns immunodetection methods and associated kits. It is proposed that antigens of the present invention may be employed to detect antibodies having reactivity therewith, or, alternatively, antibodies prepared in accordance with the present invention, may be employed to detect antigens. In general, these methods will include first obtaining a sample suspected of containing such an antigen or antibody, contacting the sample with an antibody or antigen in accordance with the present invention, as the case may be, under conditions effective to allow the antibody to form an immunocomplex with the antigen or antibody to be detected, and detecting the presence of the antigen in the sample by detecting the formation of an immunocomplex.
  • immunocomplex formation is quite well known in the art and may be achieved through the application of numerous approaches.
  • the present invention contemplates the application of ELISA, RIA, immunoblot, dotblot, indirect immunofluorescence techniques and the like.
  • immunocomplex formation will be detected through the use of a label, such as a radiolabel or an enzyme tag (such -li ⁇ as alkaline phosphatase, horseradish peroxidase, or the like) .
  • a label such as a radiolabel or an enzyme tag (such -li ⁇ as alkaline phosphatase, horseradish peroxidase, or the like) .
  • a secondary binding ligand such as a second antibody or a biotin/avidin ligand binding arrangement, as is known in the art.
  • any sample suspected of comprising either the antigen or antibody sought to be detected may be employed.
  • Exemplary samples include clinical samples obtained from a patient such as blood or serum samples, ear swabs, sputum samples, middle ear fluid or even perhaps urine samples may be employed.
  • non-clinical samples such as in the titering of antigen or antibody samples, in the selection of hybrido as, and the like.
  • kits in accordance with the present invention contemplates the preparation of kits that may be employed to detect the presence of antigens and/or antibodies in a sample.
  • kits in accordance with the present invention will include a suitable OMP antigen (i.e., either the 30 kD, 80 kD or HMWP species, or protein containing epitopes corresponding to one or more of these species) , or antibody directed against such an antigen, together with an immunodetection reagent and a means for containing the antibody or antigen and reagent.
  • the immunodetection reagent will typically comprise a label associated with the antibody or antigen, or associated with a secondary binding ligand.
  • Exemplary ligands might include a secondary antibody directed against the first antibody or antigen or a biotin or avidin (or streptavidin) ligand having an associated label.
  • the container means will generally include a vial into which the antibody, antigen or detection reagent may be placed, and preferably suitably aliquoted.
  • the kits of the present invention will also typically include a means for containing the antibody, antigen, and reagent containers in close confinement for commercial sale. Such containers may include injection or blow-molded plastic containers into which the desired vials are retained.
  • FIG. 1 Western blot analysis of M. catarrhalis proteins using as a probe monoclonal antibody 10F3, which recognizes the 80 kD OMP.
  • Lane A is a Rainbow protein molecular weight marker (M.W. 14.3 to 200 kD, Amersham);
  • Lane B is a negative control comprising a whole cell lysate of 4Bl/pBR322/RRl (4B1 is an M. catarrhalis gene encoding an unrelated protein recognized by monoclonal antibody 4B1) ;
  • Lanes C and D are whole cell lysates of 10F3/pBR322/RRl; and
  • Lane E is a blank control.
  • FIG. 1 Preliminary restriction map of pMEH120, which comprises a segment encoding the Mab 10F3-reactive 80 kD antigen.
  • FIG. 1 Preliminary restriction map of phage MEH200, which comprises a segment encoding the Mab 17C7- reactive HMWP antigen.
  • FIG. 4 Western blot analysis of M. catarrhalis proteins using as a probe monoclonal antibody 8B6, which recognizes the 30 kD OMP.
  • Lane A is a Rainbow protein molecular weight marker (M.W. 14.3 to 200 kD, Amersham); Lane B is a prestained SDS-PAGE-standard, low molecular weight (M.W. 16 to 110 kD, Bio-Rad) ; Lane C contains proteins from a phage lysate of recombinant E. coli that express the 30 kD OMP (LE392/8B6) ; Lane D is a blank control; Lane E is a negative control (phage lysate from recombinant E. coli expressing the HMWP OMP, LE392/17C7) ; and Lane F is a positive control (M. catarrhalis 035E outer membrane vesicles) .
  • FIG. 5 Western blot analysis of M. catarrhalis proteins using as a probe monoclonal antibody 17C7, which recognizes the HMWP OMP.
  • Lane A is a Rainbow protein molecular weight marker (M.W. 14.3 to 200 kD, Amersham); Lane B is a prestained SDS-PAGE-standard, low molecular weight (M.W. 16 to 110 kD, Bio-Rad) ; Lanes C, D and E contain proteins from a phage lysate of recombinant E. coli that express the HMWP OMP (LE392/17C7) ; Lane F is a blank control; Lane H is a negative control (phage lysate from recombinant E. coli expressing the 30 kD OMP, E. coli/8B6 phage lysate) ; and Lane G is a positive control (M. catarrhalis 035E outer membrane vesicles) .
  • the present invention relates to the inventors' identification of particular outer membrane proteins
  • OMPs Moraxella catarrhalis
  • These proteins appear to be cell surface-exposed in their natural state, and exhibit molecular weights of about 30 kD, 80 kD and between about 200 and 700 kD, respectively, upon SDS-PAGE.
  • Particular embodiments relate to the recombinant cloning of sequences encoding these proteins, antigenic subfragments, variants, and the like.
  • the present invention also relates to monoclonal antibodies to these M. catarrhalis OMPs that are shown to reduce the number of infecting M. catarrhalis bacteria present in localized lung infections, as demonstrated in pulmonary clearance studies using a murine model system.
  • Recombinant clones expressing one or more of the selected OMPs, and that may be used to prepare purified
  • OMP antigens as well as mutant or variant protein species in significant quantities, are included within the scope of the disclosure.
  • the selected OMP antigen, and variants thereof, are anticipated to have significant utility in diagnosing and treating M. catarrhalis infections.
  • these OMP antigens, or peptide variants may be used in immunoassays to detect M . catarrhalis or as a vaccine to treat M. catarrhalis infections.
  • plasmid pMEH300 (ATCC accession number 69049) bearing a segment encoding the 30 kD OMP antigen
  • plasmid pMEH 120 (ATCC accession number 75285) bearing a segment encoding the 80 kD OMP antigen
  • phage MEH 200 (ATCC accession number 75286) bearing a segment encoding the HMWP antigen
  • the pMEH300 plasmid can be characterized as a modified pLG338 vector in which pLG338 was digested with Xhol , and SacI linkers added.
  • This new vector contains a Moraxella catarrhalis chromosomal DNA insert of about 20 kb in size that can be excised by digestion with SacI.
  • This insert contains an M. catarrhalis gene encoding the 30 kD antigen reactive with monoclonal antibody 8B6.
  • the total vector size is therefore approximately 27 kb, with the vector comprising only about 7.3 kb.
  • pMEHlOO The gene encoding the 80 kD OMP was originally cloned in a pBR322-based recombinant plasmid, designated pMEHlOO. Subsequently, this gene was subcloned in pBluescript for sequencing analysis.
  • This new plasmid, designated pMEH120 is what was deposited with the ATCC.
  • Recombinant plasmid pMEH120 is a pBluescript II SK+ vector containing an insert of M. catarrhalis chromosomal NA approximately 4.5 kb in size, and encodes a protein of about 80 kD that is reactive with monoclonal antibody 10F3.
  • a preliminary restriction map of pMEH120 is set forth in Figure 2.
  • the ⁇ GEM-11 phage vector includes an M. catarrhalis chromosomal DNA insert of about 11 kb in size, which can be excised from the phage DNA by digestion with either Sfil or Sad .
  • a preliminary restriction map is shown in Figure 3.
  • the nucleic acid sequences which encode for the selected OMP antigen, or their variants, may be useful in hybridization or polymerase chain reaction (PCR) methodology to detect M. catarrhalis .
  • PCR polymerase chain reaction
  • the OMP antigens of the present invention are referred to, respectively, as the 30 kD, 80 kD and HMWP OMPs. These proteins have been identified by the inventors by reference to monoclonal antibodies that were selected from a battery of monoclonal antibodies against ⁇ f. catarrhalis outer membrane vesicles. These antibodies were employed as Western blot probes to identify corresponding antigens from SDS-PAGE runs of ⁇ f. catarrhalis 035E outer membrane vesicle preparations.
  • the monoclonal antibody recognizing the 30 kD OMP is termed 8B6, the antibody recognizing the 80 kD OMP is termed 10F3, and that recognizing the HMWP kD antigen has been designated 17C7 (see Figures 1, 4 and 5) .
  • each of the foregoing hybridomas have been shown to be protective against ⁇ f. catarrhalis infection in animal models.
  • hybridomas secreting the foregoing monoclonal antibodies that recognize the preferred OMP antigens have also been deposited with the ATCC under the provisions of the Budapest treaty on July 30, 1992.
  • the deposited hybridomas secrete respectively, monoclonal antibody 8B6 (ATCC accession number HB11091) , which recognizes the 30 kd OMP antigen; monoclonal antibody 10F3 (ATCC accession number HB11092) , which recognizes the 80 kD OMP antigen; and monoclonal antibody 17C7 (ATCC accession number HB11093) which recognizes the HMWP OMP antigen.
  • the present invention envisions various means for both producing and isolating the OMP antigen proteins of the present invention, ranging from isolation of purified or partially purified protein from natural sources (e.g., from ⁇ f. catarrhalis bacterial cells) , or from recombinant DNA sources (e.g., E. coli or microbial cells).
  • natural sources e.g., from ⁇ f. catarrhalis bacterial cells
  • recombinant DNA sources e.g., E. coli or microbial cells.
  • the OMP antigens of the invention, or antigenic peptides derived therefrom may be provided in essentially antigenically pure states in that they will be free of other ⁇ f. catarrhalis epitopes unrelated to the selected OMP species.
  • isolation of the OMP antigen from either natural or recombinant sources in accordance with the invention may be achieved isolating cell envelopes or outer membranes and then using a detergent- based purification scheme.
  • the desired antigen may be present in inclusion bodies.
  • Second generation proteins will typically share one or more properties in common with the full-length antigen, such as a particular antigenic/immunogenic epitopic core sequence.
  • Epitopic sequences can be provided on relatively short molecules prepared from knowledge of the peptide, or underlying DNA sequence information.
  • variant molecules may not only be derived from selected immunogenic/ antigenic regions of the protein structure, but may additionally, or alternatively, include one or more functionally equivalent amino acids selected on the basis of similarities or even differences with respect to the natural sequence.
  • epitopic core sequences are identified herein in particular aspects as hydrophilic regions of the OMP antigen. It is proposed that these regions represent those which are most likely to promote T-cell or B-cell stimulation, and, hence, elicit specific antibody production.
  • An epitopic core sequence is a relatively short stretch of amino acids that is "complementary" to, and therefore will bind, antigen binding sites on OMP-directed antibodies. Additionally or alternatively, an epitopic core sequence is one that will elicit antibodies that are cross-reactive with OMP directed antibodies. It will be understood that in the context of the present disclosure, the term
  • epitope core sequences of the present invention may be operationally defined in terms of their ability to compete with or perhaps displace the binding of the desired OMP antigen with the corresponding OMP-directed antisera.
  • the size of the polypeptide antigen is not believed to be particularly crucial, so long as it is at least large enough to carry the identified core sequence or sequences.
  • the smallest useful core sequence anticipated by the present disclosure would be on the order of about 15 amino acids in length.
  • this size will generally correspond to the smallest peptide antigens prepared in accordance with the invention.
  • the size of the antigen may be larger where desired, so long as it contains a basic epitopic core sequence.
  • the inventor proposes to identify particular hydrophilic peptidyl regions of the 30 kD, 80 kD or HMWP OMP antigen which are believed to constitute epitopic core sequences comprising particular epitopes of the protein.
  • Syntheses of epitopic sequences, or peptides which include an antigenic epitope within their sequence are readily achieved using conventional synthetic techniques such as the solid phase method (e.g., through the use of commercially available peptide synthesizer such as an Applied Biosystems Model 430A Peptide Synthesizer) .
  • Peptide antigens synthesized in this manner may then be aliquoted in predetermined amounts and stored in conventional manners, such as in aqueous solutions or, even more preferably, in a powder or lyophilized state pending use.
  • they may be readily stored in aqueous solutions for fairly long periods of time if desired, e . g.
  • aqueous storage it will generally be desirable to include agents including buffers such as Tris or phosphate buffers to maintain a pH of 7.0 to 7.5.
  • agents which will inhibit microbial growth such as sodium azide or Merthiolate.
  • For extended storage in an aqueous state it will be desirable to store the solutions at 4'C, or more preferably, frozen.
  • the peptide(s) are stored in a lyophilized or powdered state, they may be stored virtually indefinitely, e.g. , in metered aliquots that may be rehydrated with a predetermined amount of water (preferably distilled) or buffer prior to use.
  • amino acids may be substituted for other amino acids in a protein structure in order to modify or improve its antigenic or immunogenic activity (see, e.g., Kyte et al, or Hopp, U.S. patent 4,554,101, incorporated herein by reference).
  • substitution of alternative amino acids small conformational changes may be conferred upon an antigenic peptide which result in increase affinity between the antigen and the antibody binding regions.
  • amino acid substitutions in certain OMP antigenic peptides may be utilized to provide residues which may then be linked to other molecules to provide peptide-molecule conjugates which retain enough antigenicity of the starting peptide to be useful for other purposes.
  • a selected OMP peptide bound to a solid support might be constructed which would have particular advantages in diagnostic embodiments.
  • hydropathic index of amino acids in conferring interactive biologic function on a protein has been discussed generally by Kyte et al. (1982) , wherein it is found that certain amino acids may be substituted for other amino acids having a similar hydropathic index or core and still retain a similar biological activity. As displayed in the table below, amino acids are assigned a hydropathic index on the basis of their hydrophobicity and charge characteristics. It is believed that the relative hydropathic character of the amino acid determines the secondary structure of the resultant protein, which in turn defines the interaction of the protein with substrate molecules. Preferred substitutions for monitoring binding capability will generally involve amino acids having index scores within ⁇ 2 units of one another, and more preferably within ⁇ 1 unit, and even more preferably, within ⁇ 0.5 units.
  • isoleucine which has a hydropathic index of +4.5
  • an amino acid such as valine (+ 4.2) or leucine (+ 3.8).
  • lysine (- 3.9) will preferably be substituted for arginine (-4.5), and so on.
  • hydrophilicity values have been asssigned to amino acid residues: arginine (+3.0); lysine (+3.0); aspartate (+3.0 ⁇ 1); glutamate (+3.0 ⁇ 1); serine (+0.3); asparagine (+0.2); glutamine (+0.2); glycine (0) ; (0 ⁇ 1); threonine (-0.4); alanine (-0.5); histidine (-0.5); cysteine (-1.0); methionine (-1.3); valine (-1.5); leucine (-1.8); isoleucine (-1.8); tyrosine
  • amino acid can be substituted for another having a similar hydrophilicity value and still obtain a biologically equivalent, and in particular, an immunologically equivalent protein.
  • substitution of amino acids whose hydrophilicity values are within ⁇ 2 is preferred, those which are within ⁇ 1 are particularly preferred, and those within ⁇ 0.5 are even more particularly preferred.
  • amino acid substitutions are generally based on the relative similarity of R-group substituents, for example, in terms of size, electrophilic character, charge, and the like.
  • preferred substitutions which take various of the foregoing characteristics into consideration include the following:
  • Monoclonal antibodies specific for the Moraxella catarrhalis OMPs of the present invention may be prepared using conventional immunization techniques. Initially, a composition containing antigenic epitopes of the OMP, such as an outer membrane vesicle preparation, can be used to immunize an experimental animal, such as a mouse, from which a population of spleen or lymph cells are subsequently obtained. The spleen or lymph cells can then be fused with cell lines, such as human or mouse myeloma strains, to produce antibody-secreting hybridomas. These hybridomas may be isolated to obtain individual clones which can then be screened for production of antibody to the desired OMP.
  • a composition containing antigenic epitopes of the OMP such as an outer membrane vesicle preparation
  • an experimental animal such as a mouse
  • the spleen or lymph cells can then be fused with cell lines, such as human or mouse myeloma strains, to produce antibody-secreting hybridomas. These hybrido
  • the present invention utilizes outer membrane fragments from ⁇ f. catarrhalis to induce an immune response in experimental animals.
  • spleen cells are removed and fused, using a standard fusion protocol (see, e.g.. The Cold Spring Harbor Manual for Hybridoma Development, incorporated herein by reference) with plasmacytoma cells to produce hybridomas secreting monoclonal antibodies against outer membrane proteins.
  • Hybridomas which produce monoclonal antibodies to the selected OMP are identified using standard techniques, such as ELISA and Western blot methods.
  • Hybridoma clones can then be cultured in liquid media and the culture supernatants purified to provide the OMP-specific monoclonal antibodies.
  • monoclonal antibodies to the desired OMP antigen of ⁇ f. catarrhalis can be used in both the diagnosis and treatment of ⁇ f. catarrhalis infections.
  • the monoclonal antibodies of the present invention will find useful application in standard immunochemical procedures, such as ELISA and Western blot methods, as well as other procedure which may utilize antibody specific to OMP epitopes.
  • These OMP-specific monoclonal antibodies are anticipated to be useful various ways for the treatment of Af. catarrhalis infections through, for example, their application in passive immunization procedures.
  • monoclonal antibodies specific to the particular OMP may be utilized in other useful applications. For example, their use in immunoabsorbent protocols may be useful in purifying native or recombinant OMP species or variants thereof.
  • OMP antigens of the invention have a significant protective effect against ⁇ f. catarrhalis infection.
  • the present inventors have shown that passive immunization with monoclonal antibodies specific for these OMPs significantly reduce the numbers of Af. catarrhalis organisms following a bolus injection of bacteria. This demonstrates that these OMP antigens may be employed in making gammaglobulin preparations for use in passive immunization against disorders associated with ⁇ f. catarrhalis infections, or used directly as vaccine components.
  • gammaglobulin preparations one may desire to prepare monoclonal antibodies, preferably human or humanized hybridomas. Alternatively, it is proposed that one may desire to use globulin fractions from hyperimmunized individuals.
  • the present invention also involves isolating ⁇ f. catarrhalis OMP genes, or sequence variants, incorporating DNA segments encoding the 30 kD, 80 kD or HMWP OMP gene into a suitable vector, and transforming a suitable host, such that recombinant proteins, or variants thereof, are expressed.
  • OMP gene sequences from any suitable source that includes appropriate coding sequences, such as any M. catarrhalis subspecies or isolate that expresses the desired OMP.
  • Such sources may be readily identified by immunological screening with monoclonal antibodies to the selected OMP.
  • the preferred application of the present invention to the isolation and use of OMP-encoding DNA involves generally the steps of (1) isolation of Moraxella genomic DNA; (2) partial restriction enzyme digestion of the genomic DNA with an enzyme such as PstI, (the selected restriction enzyme is not crucial) to provide DNA having an average length of, e.g., 6 to 23 kb; (3) ligation of the partially digested DNA into a selected site within a selected vector, such as pBR322 (again, other plasmid or phage vectors may be used at this step, as desired) ; (4) transformation, transfection or electroporation of suitable host cells, e.g., E.
  • genomic DNA from Moraxella catarrhalis strain 035E was isolated from bacteria through the use of SDS, ribonuclease and proteinase K treatment, phenol/chloroform/isoamyl alchohol extraction and ethanol precipitation. Conditions were determined for achieving an appropriate partial restriction enzyme digestion, such as would provide fragments on the order of 6-23 kb in length, using a restriction enzyme, such as Pstl. After size fractionation, the partially digested Moraxella DNA fragments of the selected size range were ligated with fully digested vector, such as pBR322, which was fully digested with Pstl to generate compatible sites for ligation with the genomic DNA fragments.
  • the recombinant vectors are then used to transform a suitable host, such as E. coli RRl, to produce a recombinant library having members that express ⁇ f. catarrhalis protein species encoded by the DNA fragment inserts.
  • the recombinant microbial clones are cultivated, preferably on the surface of a nutrient agar, to form visible colonies. Those colonies expressing surface-exposed ⁇ f. catarrhalis outer membrane proteins are then identified using monoclonal antibodies to ⁇ f. catarrhalis OMPs in a colony blot radioimmunoassay.
  • Recombinant E. coli clones expressing proteins having epitopes reactive with anti-OMP antibodies may then be cultured in desired quantities.
  • E. coli strain RRl is particularly useful.
  • Other microbial strains which may be used include E. coli strains such as E. coli LE392, E. coli B, and E. coli X 1776 (ATCC No. 31537) . These examples are, of course, intended to be illustrative rather than limiting.
  • Prokaryotes are also preferred for expression.
  • the aforementioned strains, as well as E. coli W3110 (F-, lambda-, prototrophic, ATCC No. 273325) , bacilli such as Bacillus subtilis , or other enterobacteriaceae such as Salmonella typhimurium or Serratia marcescens , and various Pseudomonas species may be used.
  • plasmid vectors containing replicon and control sequences which are derived from species compatible with the host cell are used in connection with these hosts.
  • the vector ordinarily carries a replication site, as well as marking sequences which are capable of providing phenotypic selection in transformed cells.
  • E. coli is typically transformed using pBR322, a plasmid derived from an E. coli species (Bolivar et al . , 1977) .
  • pBR322 contains genes for ampicillin and tetracycline resistance and thus provides easy means for identifying transformed cells.
  • the pBR plasmid, or other microbial plasmid or phage must also contain, or be modified to contain, promoters which can be used by the microbial organism for expression of its own proteins.
  • phage vectors containing replicon and control sequences that are compatible with the host microorganism can be used as a transforming vector in connection with these hosts.
  • the phage lambda GEMTM-11 may be utilized in making recombinant phage vector which can be used to transform host cells, such as E. coli LE392.
  • promoters most commonly used in recombinant DNA construction include the B-lactamase (penicillinase) and lactose promoter systems (Chang et al . , 1978; Itakura et al . , 1977; Goeddel et al . , 1979) and a tryptophan (trp) promoter system (Goeddel et al . , 1980; EPO Appl. Publ. No. 0036776). While these are the most commonly used, other microbial promoters have been discovered and utilized, and details concerning their nucleotide sequences have been published, enabling a skilled worker to ligate them functionally with plasmid vectors (EPO Appl. Publ. No. 0036776).
  • eukaryotic microbes such as yeast cultures may also be used.
  • Saccharomyces cerevisiae or common baker's yeast is the most commonly used among eukaryotic microorganisms, although a number of other strains are commonly available.
  • the plasmid YRp7 for example, is commonly used (Stinchcomb et al . , 1979; Kingsman et al . , 1979; Tschemper et al . , 1980).
  • This plasmid already contains the trpl gene which provides a selection marker for a mutant strain of yeast lacking the ability to grow in tryptophan, for example ATCC No. 44076 or PEP4-1 (Jones, 1977) .
  • the presence of the trpl lesion as a characteristic of the yeast host cell genome then provides an effective environment for detecting transformation by growth in the absence of tryptophan.
  • Suitable promoting sequences in yeast vectors include the promoters for 3-phosphoglycerate kinase (Hitzeman et al . , 1980) or other glycolytic enzymes (Hess et al . , 1968; Holland et al . , 1978), such as enolase, glyceraldehyde-3-phosphate dehydrogenase, hexokinase, pyruvate decarboxylase, phosphofructokinase, glucose-6- phosphate isomerase, 3-phosphoglycerate mutase, pyruvate kinase, triosephosphate isomerase, phosphoglucose isomerase, and glucokinase.
  • 3-phosphoglycerate kinase Hitzeman et al . , 1980
  • other glycolytic enzymes Hess et al . , 1968; Holland et al . , 1978
  • enolase
  • the termination sequences associated with these genes are also ligated into the expression vector 3' of the sequence desired to be expressed to provide polyadenylation of the mRNA and termination.
  • Other promoters which have the additional advantage of transcription controlled by growth conditions are the promoter region for alcohol dehydrogenase 2 , isocytochrome C, acid phosphatase, degradative enzymes associated with nitrogen metabolism, and the aforementioned glyceraldehyde-3-phosphate dehydrogenase, and enzymes responsible for maltose and galactose utilization.
  • Any plasmid vector containing a yeast- compatible promoter, origin of replication and termination sequences is suitable.
  • cultures of cells derived from multicellular organisms may also be used as hosts.
  • any such cell culture is workable, whether from vertebrate or invertebrate culture.
  • vertebrate cells have been greatest in vertebrate cells, and propagation of vertebrate cells in culture (tissue culture) has become a routine procedure in recent years (Tissue Culture, 1973) .
  • useful host cell lines are VERO and HeLa cells, Chinese hamster ovary (CHO) cell lines, and W138, BHK, COS-7, 293 and MDCK cell lines.
  • Expression vectors for such cells ordinarily include (if necessary) an origin of replication, a promoter located in front of the gene to be expressed, along with any necessary ribosome binding sites, RNA splice sites, polyadenylation site, and transcriptional terminator sequences.
  • control functions on the expression vectors are often provided by viral material.
  • promoters are derived from polyoma, Adenovirus 2, and most frequently Simian Virus 40 (SV40) .
  • the early and late promoters of SV40 virus are particularly useful because both are obtained easily from the virus as a fragment which also contains the SV40 viral origin of replication (Fiers et al . , 1978). Smaller or larger SV40 fragments may also be used, provided there is included the approximately 250 bp sequence extending from the Hind III site toward the Bgl I site located in the viral origin of replication.
  • promoter or control sequences normally associated with the desired gene sequence are compatible with the host cell systems.
  • origin of replication may be provided either by construction of the vector to include an exogenous origin, such as may be derived from SV40 or other viral (e.g., Polyo a, Adeno, VSV, BPV) source, or may be provided by the host cell chromosomal replication mechanism. If the vector is integrated into the host cell chromosome, the latter is often sufficient.
  • exogenous origin such as may be derived from SV40 or other viral (e.g., Polyo a, Adeno, VSV, BPV) source, or may be provided by the host cell chromosomal replication mechanism. If the vector is integrated into the host cell chromosome, the latter is often sufficient.
  • hydrophilic peptidyl regions of the OMP antigen may be identified which are likely to constitute epitopic core sequences, comprising particular epitopes of the protein, as well as biologically functional equivalents of the foregoing peptides, as explained in more detail below.
  • Site-specific mutagenesis is a technique useful in the preparation of individual peptides, or biologically functional equivalent proteins or peptides, derived from the OMP antigen sequence, through specific mutagenesis of the underlying DNA.
  • the technique further provides a ready ability to prepare and test sequence variants, for example, incorporating one or more of the foregoing considerations, by introducing one or more nucleotide sequence changes into the DNA.
  • Site-specific mutagenesis allows the production of mutants through the use of specific oligonucleotide sequences which encode the DNA sequence of the desired mutation, as well as a sufficient number of adjacent nucleotides, to provide a primer sequence of sufficient size and sequence complexity to form a stable duplex on both sides of the deletion junction being traversed.
  • a primer of about 17 to 25 nucleotides in length is preferred, with about 5 to 10 residues on both sides of the junction of the sequence being altered.
  • the technique of site-specific mutagenesis is well known in the art as exemplified by publications (Adelman et al . , 1983). As will be appreciated, the technique typically employs a phage vector which exists in both a single stranded and double stranded form.
  • Typical vectors useful in site-directed mutagenesis include vectors such as the M13 phage (Messing et al . , 1981). These phage are readily commercially available and their use is generally well known to those skilled in the art.
  • site-directed mutagenesis in accordance herewith is performed by first obtaining a single- stranded vector which includes within its sequence a DNA sequence which encodes the OMP antigen.
  • An oligonucleotide primer bearing the desired mutated sequence is prepared, generally synthetically, for example by the method of Crea et al . (1978).
  • This primer is then annealed with the single-stranded vector, and subjected to DNA polymerizing enzymes such as E. coli polymerase I Klenow fragment, in order to complete the synthesis of the mutation-bearing strand.
  • DNA polymerizing enzymes such as E. coli polymerase I Klenow fragment
  • OMP gene using site-directed mutagenesis is provided as a means of producing potentially useful OMP species and is not meant to be limiting as there are other ways in which sequence variants of the OMP may be obtained.
  • recombinant vectors encoding the desired OMP gene may be treated with mutagenic agents to obtain sequence variants (see, e.g., a method described by Eichenlaub, 1979) for the mutagenesis of plasmid DNA using hydroxylamine.
  • the DNA sequence information provided by the present disclosure allows for the preparation of relatively short DNA (or RNA) sequences having the ability to specifically hybridize to gene sequences of the selected OMP antigen gene.
  • nucleic acid probes of an appropriate length are prepared based on a consideration of the natural sequence or derived from flanking regions of the OMP gene, such as regions downstream of the gene as found in plasmid pBR322.
  • the ability of such nucleic acid probes to specifically hybridize to OMP gene sequences lend them particular utility in a variety of embodiments.
  • the probes can be used in a variety of diagnostic as... ays for detecting the presence of pathogenic organisms in a given sample.
  • other uses are envisioned, including the use of the sequence information for the preparation of mutant species primers, or primers for use in preparing other genetic constructions.
  • the preferred nucleic acid sequence employed for hybridization studies or assays includes sequences that are complementary to at least a 10 to 20, or so, nucleotide stretch of the sequence.
  • a size of at least 10 nucleotides in length helps to ensure that the fragment will be of sufficient length to form a duplex molecule that is both stable and selective.
  • Molecules having complementary sequences over stretches greater than 10 bases in length are generally preferred, though, in order to increase stability and selectivity of the hybrid, and thereby improve the quality and degree of specific hybrid molecules obtained.
  • Such fragments may be readily prepared by, for example, directly synthesizing the fragment by chemical means, by application of nucleic acid reproduction technology, such as the PCR technology of U.S. Patent 4,603,102, or by introducing selected sequences into recombinant vectors for recombinant production.
  • the present invention will find particular utility as the basis for diagnostic hybridization assays for detecting OMP-specific RNA or DNA in clinical samples.
  • Exemplary clinical samples that can be used in the diagnosis of infections are thus any samples which could possibly include Moraxella nucleic acid, including middle ear fluid, sputum, bronchoalveolar fluid, amniotic fluid or the like.
  • a variety of hybridization techniques and systems are known which can be used in connection with the hybridization aspects of the invention, including diagnostic assays such as those described in Falkow et al. , U.S. Patent 4,358,535.
  • nucleotide sequences of the invention are important for their ability to selectively form duplex molecules with complementary stretches of the corresponding OMP genes.
  • one will desire to employ varying conditions of hybridization to achieve varying degrees of selectivity of the probe toward the target sequence.
  • relatively stringent conditions for applications requiring a high degree of selectivity, one will typically desire to employ relatively low salt and/or high temperature conditions, such as provided by 0.02M-0.15M NaCl at temperatures of 50 ⁇ C to 70°C. These conditions are particularly selective, and tolerate little, if any, mismatch between the probe and the template or target strand.
  • mutant clone colonies growing on solid media which contain variants of the OMP sequence could be identified on duplicate filters using hybridization conditions and methods, such as those used in colony blot assays, to only obtain hybridization between probes containing sequence variants and nucleic acid sequence variants contained in specific colonies.
  • small hybridization probes containing short variant sequences of the OMP gene may be utilized to identify those clones growing on solid media which contain sequence variants of the entire OMP gene. These clones can then be grown to obtain desired quantities of the variant OMP nucleic acid sequences or the corresponding OMP antigen.
  • nucleic acid sequences of the present invention are used in combination with an appropriate means, such as a label, for determining hybridization.
  • appropriate indicator means include radioactive, enzymatic or other ligands, such as avidin/biotin, which are capable of giving a detectable signal.
  • an enzyme tag such as urease, alkaline phosphatase or peroxidase, instead of radioactive or other environmental undesirable reagents.
  • colorimetric indicator substrates are known which can be employed to provide a means visible to the human eye or spectrophoto- metrically, to identify specific hybridization with pathogen nucleic acid-containing samples.
  • the hybridization probes described herein will be useful both as reagents in solution hybridizations as well as in embodiments employing a solid phase.
  • the test DNA (or RNA) from suspected clinical samples such as exudates, body fluids (e.g., amniotic fluid, middle ear effusion, bronchoalveolar lavage fluid) or even tissues, is adsorbed or otherwise affixed to a selected matrix or surface. This fixed, single-stranded nucleic acid is then subjected to specific hybridization with selected probes under desired conditions.
  • the selected conditions will depend on the particular circumstances based on the particular criteria required (depending, for example, on the G+C contents, type of target nucleic acid, source of nucleic acid, size of hybridization probe, etc.). Following washing of the hybridized surface so as to remove nonspecifically bound probe molecules, specific hybridization is detected, or even quantified, by means of the label.
  • OMP sequences or variants thereof may be used to provide highly specific and sensitive detection of ⁇ f. catarrhalis when used as reagents in polymerase chain reaction (PCR) assays.
  • PCR polymerase chain reaction
  • various portions of the OMP sequence as oligonucleo-tide probes for the PCR amplification of a defined portion of OMP nucleic acid in a sample.
  • the amplified portion of the OMP sequence may then be detected by hybridization with a hybridization probe containing a complementary sequence. In this manner, extremely small concentrations of ⁇ f.
  • catarrhalis nucleic acid may detected in a sample utilizing OMP sequences.
  • OMP sequences may be utilized in PCR formats for the in vitro preparation of desired quantities of selected portions of the OMP gene. By amplifying selected gene portions of a selected OMP gene and then incorporating those portions into vectors, one can also prepare recombinant clones which express OMP variants, including subfragments of the OMP antigen. In this manner, peptides carrying antigen epitopes of the outer membrane protein may be prepared and utilized for various purposes.
  • OMP peptides of the invention will find utility as immunogens, e.g., in connection with vaccine development, or as antigens in immunoassays for the detection of anti-OMP antigen- reactive antibodies.
  • immunoassays in their most simple and direct sense, preferred immunoassays of the invention include the various types of enzyme linked immunosorbent assays (ELISAs) known to the art.
  • ELISAs enzyme linked immunosorbent assays
  • OMP peptides is not limited to such assays, and that other useful embodiments include RIAs and other non-enzyme linked antibody binding assays or procedures.
  • peptides incorporating OMP antigen sequences are immobilized onto a selected surface, preferably a surface exhibiting a protein affinity such as the wells of a polystyrene microtiter plate.
  • a selected surface preferably a surface exhibiting a protein affinity
  • a nonspecific protein such as bovine serum albumin (BSA) or casein onto the well that is known to be antigenically neutral with regard to the test antisera.
  • BSA bovine serum albumin
  • the immobilizing surface is contacted with the antisera or clinical or biological extract to be tested in a manner conducive to immune complex (antigen/antibody) formation.
  • Such conditions preferably include diluting the antisera with diluents such as BSA, bovine gamma globulin (BGG) and phosphate buffered saline (PBS)/Tween. These added agents also tend to assist in the reduction of nonspecific background.
  • BSA bovine gamma globulin
  • PBS phosphate buffered saline
  • the layered antisera is then allowed to incubate for from 2 to 4 hours, at temperatures preferably on the order of 25° to 27"C. Following incubation, the antisera-contacted surface is washed so as to remove non-immunocomplexed material.
  • a preferred washing procedure includes washing with a solution such as PBS/Tween, or borate buffer.
  • the occurrence and even amount of immunocomplex formation may be determined by subjecting same to a second antibody having specificity for the first.
  • the second antibody will preferably be an antibody having specificity in general for human IgG.
  • the second antibody will preferably have an associated enzyme that will generate a color development upon incubating with an appropriate chromogenic substrate.
  • a urease or peroxidase-conjugated anti-human IgG for a period of time and under conditions which favor the development of immunocomplex formation (e.g., incubation for 2 hours at room temperature in a PBS-containing solution such as PBS-Tween) .
  • the amount of label is quantified by incubation with a chromogenic substrate such as urea and bromocresol purple or 2,2'-azino-di-(3-ethyl-benzthiazoline-6- sulfonic acid [ABTS] and H 2 0 2 , in the case of peroxidase as the enzyme label. Quantification is then achieved by measuring the degree of color generation, e.g., using a visible spectra spectrophotometer.
  • a chromogenic substrate such as urea and bromocresol purple or 2,2'-azino-di-(3-ethyl-benzthiazoline-6- sulfonic acid [ABTS] and H 2 0 2 , in the case of peroxidase as the enzyme label.
  • Quantification is then achieved by measuring the degree of color generation, e.g., using a visible spectra spectrophotometer.
  • Immunogenic compositions proposed to be suitable for use as a vaccine, may be prepared most readily directly from immunogenic OMP proteins and/or peptides prepared in a manner disclosed herein.
  • the antigenic material is extensively dialyzed to remove undesired small molecular weight molecules and/or lyophilized for more ready formulation into a desired vehicle.
  • vaccines which contain peptide sequences as active ingredients is generally well understood in the art, as exemplified by U.S. Patents 4,608,251; 4,601,903; 4,599,231; 4,599,230; 4,596,792; and 4.578,770, all incorporated herein by reference.
  • Such vaccines are prepared as injectables. Either as liquid solutions or suspensions: solid forms suitable for solution in, or suspension in, liquid prior to injection may also be prepared. The preparation may also be emulsified.
  • the active immunogenic ingredient is often mixed with excipients which are pharmaceutically acceptable and compatible with the active ingredient.
  • Suitable excipients are, for example, water, saline, dextrose, glycerol, ethanol, or the like and combinations thereof.
  • the vaccine may contain minor amounts of auxiliary substances such as wetting or emulsifying agents, pH buffering agents, or adjuvants which enhance the effectiveness of the vaccines.
  • the vaccines are conventionally administered parenterally, by injection, for example, either subcutaneously or intramuscularly.
  • Additional formulations which are suitable for other modes of administration include suppositories and, in some cases, oral formulations.
  • suppositories traditional binders and carriers may include, for example, polyalkalene glycols or triglycerides: such suppositories may be formed from mixtures containing the active ingredient in the range of 0.5% to 10%, preferably 1-2%.
  • Oral formulations include such normally employed excipients as, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate and the like. These compositions take the form of solutions, suspensions, tablets, pills, capsules, sustained release formulations or powders and contain 10-95% of active ingredient, preferably 25-70%.
  • the proteins may be formulated into the vaccine as neutral or salt forms.
  • Pharmaceutically acceptable salts include the acid addition salts (formed with the free amino groups of the peptide) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups may also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, 2-ethylamino ethanol, histidine, procaine, and the like.
  • the vaccines are administered in a manner compatible with the dosage formulation, and in such amount as will be therapeutically effective and immunogenic.
  • the quantity to be administered depends on the subject to be treated, including, e.g.. the capacity of the individual's immune system to synthesize antibodies, and the degree of protection desired.
  • Precise amounts of active ingredient required to be administered depend on the judgment of the practitioner. However, suitable dosage ranges are of the order of several hundred micrograms active ingredient per vaccination. Suitable regimes for initial administration and booster shots are also variable, but are typified by an initial administration followed by subsequent inoculations or other administrations.
  • Any of the conventional methods for administration of a vaccine are applicable. These are believed to include oral application on a solid physiologically acceptable base or in a physiologically acceptable dispersion, parenterally, by injection or the like.
  • the dosage of the vaccine will depend on the route of administration and will vary according to the size of the host.
  • Various methods of achieving adjuvant effect for the vaccine includes use of agents such as aluminum hydroxide or phosphate (alum), commonly used as 0.05 to 0.1 percent solution in phosphate buffered saline, admixture with synthetic polymers of sugars (Carbopol) used as 0.25 percent solution, aggregation of the protein in the vaccine by heat treatment with temperatures ranging between 70 * to 101"C for 30 second to 2 minute periods respectively.
  • agents such as aluminum hydroxide or phosphate (alum), commonly used as 0.05 to 0.1 percent solution in phosphate buffered saline, admixture with synthetic polymers of sugars (Carbopol) used as 0.25 percent solution, aggregation of the protein in the vaccine by heat treatment with temperatures ranging between 70 * to 101"C for 30 second to 2 minute periods respectively.
  • Fab pepsin treated antibodies to albumin
  • mixture with bacterial cells such as C ⁇ parvum or endotoxins or lipopolysaccharide components of gram-negative bacteria
  • emulsion in physiologically acceptable oil vehicles such as mannide mono-oleate (Aracel A) or emulsion with 20 percent solution of a perfluorocarbon (Fluosol-DA) used as a block substitute
  • oil vehicles such as mannide mono-oleate (Aracel A) or emulsion with 20 percent solution of a perfluorocarbon (Fluosol-DA) used as a block substitute
  • the vaccine will be desirable to have multiple administrations of the vaccine, usually not exceeding six vaccinations, more usually not exceeding four vaccinations and preferably one or more, usually at least about three vaccinations.
  • the vaccinations will normally be at from two to twelve week intervals, more usually from three to five week intervals. Periodic boosters at intervals of 1-5 years, usually three years, will be desirable to maintain protective levels of the antibodies.
  • the course of the immunization may be followed by assays for antibodies for the supernatant antigens.
  • the assays may be performed by labeling with conventional labels, such as radionuclides, enzymes, fluorescers, and the like. These techniques are well known and may be found in a wide variety of patents, such as U.S. Patent Nos. 3,791,932; 4,174,384 and 3,949,064, as illustrative of these types of assays.
  • outer membrane fragments from ⁇ f. catarrhalis strain 035E were prepared as an immunogen.
  • ⁇ f. catarrhalis strain 035E cells were grown on agar plates using brain heart infusion broth. Plates were incubated at 37' C in a candle extinction jar. Outer membrane fragments were subsequently prepared from these cells by the EDTA-based extraction procedure of Murphy et al . , Microb . Path . , 1989.
  • OMP antigen may be purified using the following general procedure.
  • Cell envelopes will be prepared by sonication or outer membrane fragments will be extracted by EDTA- based treatment of whole ⁇ f. catarrhalis cells.
  • These membranes will be treated with ionic or non-ionic detergents to release the desired proteins which can then be purified by using conventional column chromatography or by immunoaffinity techniques.
  • the present example illustrates the steps employed by the inventors in reducing certain aspects of the invention to practice.
  • this example relates to the generation and identification of hybridomas that produce monoclonal antibodies to the 30 kD, 80 kD or HMWP OMP antigen.
  • hybridomas secreting monoclonal antibodies to surface-exposed OMP antigens from Af. catarrhalis were identified, those determined to produce antibody to these OMP antigens were selected and cultured to produce antibody for use in other studies, such as those involving pulmonary clearance of ⁇ f. catarrhalis.
  • BALB/c mice were immunized by intraperitoneal injection with outer membrane fragments of ⁇ f. catarrhalis strain 035E prepared by the EDTA-based extraction procedure.
  • mice were immunized with 50-100 ⁇ g protein in 0.1 ml of Freund's complete adjuvant. One month later, the animals were boosted with an identical quantity of this same protein preparation in incomplete Freund's adjuvant. Three weeks later, the mice were given an intravenous injection (into the tail vein) with 50 ⁇ g protein of the same membrane preparation suspended in PBS.
  • SP 2/0 -Agl4 plasmacytoma cells were employed. These cells were maintained in DMEM (Dulbecco's Modified Eagle Medium)/ Penicillin-Strepto ycin-Glutamine with 15% fetal bovine serum, 1% Fungizone and 8-azaguanine. Two weeks prior to the fusion, some of the cells were split into media with 1% Fungizone but lacking 8-azaguanine. These cells were maintained for 10 days at a density of no greater than 1-2 x 10 5 /ml. Beginning three days before the fusion, SP ⁇ Q cells were subcultured every 24 hours and maintained at an approximate density of 2-3 x l0 5 /ml. Three days before the fusion, the mice were boosted intravenously with about 50 ⁇ g of protein immunogen. On the day of the fusion, two mice were sacrificed by cervical dislocation.
  • DMEM Dynamic Eagle Medium
  • Penicillin-Strepto ycin-Glutamine with 15% fetal bovine serum
  • the spleens were removed aseptically and macerated. Spleen cells were collected in 10 mis of DMEM-HY media (60 ml NCTC-109, 6 tubes hypoxanthine-thymidine-glycine stock soln. , 6 tubes oxaloacetic acid-bovine insulin stock soln., 12 ml penicillin-streptomycin-glutamine, 2.7 ml lOOmM Na pyruvate, and 508 ml DMEM) . At room temperature, SP 2/0 cells and spleen cells were collected by centrifugation at 170 x g for 11 min. in their respective tubes. SP 20 cells and spleen cells were each resuspended in a total of 5 mis of DMEM-HY media.
  • hypoxanthine-thymidine-glycine stock solution was prepared by adding 136 mg hypoxanthine to 100 ml 0.1 M HCl, 38.7 mg thymidine to 100 ml H 2 0, and 2.3 mg glycine to 20 ml H 2 0. These solutions were dissolved separately, combined and then aliquoted into 2.2 ml volumes.
  • the oxaloacetic acid-bovine insulin stock solution was prepared by dissolving 80.3 mg bovine insulin in 100 ml H 2 0, adding 1.32 gm oxaloacetic acid and aliquoting into l ml. volumes.
  • Spleen cells were then diluted to 2 x 10 8 cells/5 mis and the SP ⁇ o cells was diluted to 2 x 10 7 cells/5 mis.
  • the ratio of spleen to SP 2/0 cells was 10:1.
  • Spleen cells were then mixed with SP 2/0 cells in a ratio of 1:1.
  • the spleen-SPj / o mixture was then treated with 3 mis of 50% PEG/DMEM-HY media for 35 sec.
  • Fused spleen-SP j/ o cells were washed immediately with DMEM-HY and incubated in 30% HY.-HIFCS (35 ml DMEM-HY, 15 ml FBS, filter) for 24 hours at 37°C.
  • Each Mab identified as being reactive with ⁇ f. catarrhalis in Western blot analysis was used in the indirect antibody accessibility assay to determine if these Mabs were reactive with surface-exposed determinants of this organism.
  • the antibody accessibility assay performed was described by Patrick et al., 1987.
  • Mab 10F3 which reacted with an antigen with an apparent MW of approximately 80,000 in Western blot analysis, was shown to bind to the surface of whole cells of strain 035E. This Mab reacted with 4 of 10 different ⁇ f. catarrhalis strains tested in colony blot-RIA analysis by the method of Gulig et al., 1987.
  • Mab 17C7 reacted with two different size bands in Western blot analysis. This Mab reacted with a band near the top of the gel that migrated in a diffuse form and sometimes with a second band that migrated with an apparent MW of between about 200 and about 700 kD. For the purpose of clarity, the Mab will be defined as being reactive with the "HMWP" antigen. This Mab bound to the surface of strain 035E and reacted with all ten different ⁇ f. catarrhalis strains tested in the colony blot RIA. Mab 8B6 reacted with an antigen with an apparent MW of approximately 30,000 in Western blot analysis. This Mab was also reactive with the surface of strain 035E and reacted with all ten different ⁇ f. catarrhalis strains tested in the colony blot-RIA.
  • the present example illustrates steps employed by the inventors in reducing certain aspects of the invention to practice.
  • This example demonstrates the ability of monoclonal antibodies to the 30 kD, 80 kD and HMWP OMPs to enhance pulmonary clearance of ⁇ f. catarrhalis using a murine model system.
  • antibodies to the 30 kD, 80 kD or HMWP OMP may be useful for passive immunization and that vaccines comprising these OMPs are likely to provide active immunity against ⁇ f. catarrhalis infections.
  • mice Eighteen hours prior to bacterial challenge, groups of 5 mice were passively immunized by intravenous administration of monoclonal antibody 17C7, 8B6 or 10F3. Control animals were immunized with an irrelevant antibody, 2H11, which was directed against an outer membrane protein of Haemophilus ducreyi . Each animal received an equivalent amount of purified antibody corresponding to 150 ⁇ g of total protein.
  • mice were anaesthetized by intramuscular injection of 2 mg of keta ine HCL (Fort Dodge Lab, Fort Dodge, IA) and 0.2 mg of acepromazine maleate (Fort Dodge Lab). After tracheal exposure each animal was intubated transorally with a 20 gauge intravenous catheter which was advanced until it could be visualized through the translucent tracheal wall. A PE-10 polyethylene tube containing 5 ⁇ l of bacterial suspension was then passed through the catheter into the lung where the bacteria were deposited with 150 ⁇ l of air. This technique delivered the inoculum to a localized, peripheral segment of the lung. In all experiments, mice were challenged with ⁇ f. catarrhalis strain 035E.
  • mice were sacrificed by intraperitoneal injection of 0.75 mg of sodium pentobarbital (Abbott Labs, Chicago, IL) immediately after inoculation (O h), to determine bacterial deposition in the lungs.
  • experimental (17C7-, 8B6- or 10F3-immunized) and control (2H11 immunized) groups were sacrificed, and the number of viable bacteria remaining in the lungs was determined as follows: the lungs from each animal were removed aseptically and homogenized in 2 ml of sterile BHI broth in a tissue homogenizer followed by grinding in a tissue grinder until smooth.
  • the homogenate was serially diluted in BHI broth, plated on BHI agar and incubated at 37°C in an air incubator with a 5% C0 2 atmosphere for 24 h. Clearance of ⁇ f. catarrhalis from the lungs is expressed as the percentage of colony forming units (cfu) remaining in the lung at each time point compared with the mean cfu of bacteria present at 0 h in the same experiment. RESULTS
  • the mean number of viable bacteria remaining in the lungs of immunized and control mice after bolus deposition of 0.98 x 10 5 to 2.0 x 10 5 cfu of ⁇ f. catarrhalis 035E was determined and expressed as a percentage of the initial inoculum.
  • the present example illustrates steps employed by the inventor in cloning the gene encoding for the 80 kD OMP from ⁇ f. catarrhalis .
  • the present example discloses one or more preferred recombinant E. coli clones, expressing the 80 kD OMP antigen, isolated by the following procedures.
  • ⁇ f. catarrhalis strain 035E was used as a representative Moraxella pathogen in this study. Genomic DNA from ⁇ f. catarrhalis strain 035E was extracted and purified as follows. Af. catarrhalis cells (approximately 2 gms wet weight) were scraped from agar plates and resuspended in 20 mis. PBS. To this suspension was added 3.2 ml 10% (w/v) SDS and 1 ml RNase (10 mg/ml). This mixture was incubated at 37°C, then 3 mg proteinase K added, followed by further incubation at 55°C overnight.
  • the incubated mixture was then extracted once with phenol, twice with phenol:chloroform:isoamyl alcohol, and three times with chloroform:isoamyl alcohol.
  • the resulting DNA was then precipitated with two volumes of absolute ethanol, and collected with a Pasteur pipet.
  • genomic DNA was achieved by incubating 100 ⁇ g portions of ⁇ f. catarrhalis genomic DNA with varying amounts of the restriction enzyme Pstl in a reaction volume of about 1.5 ml. at 37 * C. for 1 hr.
  • the partially digested genomic DNA was then size fractionated by sucrose density gradient centrifugation. Fractions containing DNA fragments from about 6 kb to 23 kb in length were selected and dialyzed to obtain purified genomic DNA fragments for ligation with pBR322.
  • the plasmid vector pBR322 was fully digested with Pstl by incubating 15 ⁇ g portions of pBR322 with 50 units of Pstl in a 100 ⁇ l reaction volume at 37°C. for 18 hrs. Ligation of the purified DNA fragments into the Pstl- digested pBR322 vector was accomplished by incubating 300 ng of the purified DNA fragments and Pstl-digested pBR322 together with ATP and T4 DNA ligase under conditions described by Maniatis et al . (1982). After ligation, the DNA was diluted 1:5 with 10 mM TRis-HCl (pH 8.0) and was used to transform E. coli RRl made competent by the CaCl 2 method.
  • a colony blot RIA was then carried out as described by Gulig et al . (1987) with monoclonal antibody 10F3 as the primary antibody.
  • Clones which reacted with monoclonal antibody 10F3 in the colony blot RIA were cultured using LB medium containing the antibiotic tetracycline (15 ⁇ g/ml) .
  • Whole cell lysates of recombinant E. coli RRl expressing ⁇ f. catarrhalis OMP antigens were prepared as described by Patrick et al . , 1987. Briefly, portions of these whole- cell lysates were subjected to SDS-PAGE as described in Gulig et al . , 1987, and then stained with Coomassie blue or transferred to nitrocellulose for Western blot analysis.
  • DNA was isolated and purified from strain 035E, and 100 ⁇ g samples were partially digested with Sau3A (Promega Biotech) at room temperature, as described above for Pstl.
  • the digested DNA was size-fractionated in sucrose density gradients and fragments of DNA 15 kb and larger were collected for use in library construction. These DNA fragments (1 ⁇ g) were filled in using the Klenow procedure (Promega) at 14°C for 90 min. These fragments were then cleaned by standard procedures and ligated onto the phage DNA arms and packaged using the protocol and reagents supplied by Promega in the LambdaGEM - 11* Xho I Half-Site Arms Cloning System, except that T4 DNA ligase from BRL was used. After packaging, the phage-based library was titered using E. coli LE392. This genomic library contained 50,000 recombinant clones.
  • Liquid lysate cultures of these recombinant phage were prepared by the standard methods described in Current Protocols in Molecular Biology. The DNA was extracted using standard methods. Phage harvested from liquid lysates were heated at 100°C for 3 min. in standard SDS digestion buffer and then used for SDS-PAGE and Western blot analysis to confirm that these recombinant phage were expressing the appropriate ⁇ f. catarrhalis antigens.
  • the recombinant phage reactive with Mab 17C7 designated MEH200, were found to comprise DNA coding for the HMWP.
  • a preliminary restriction map of MEH200 is shown in Figure 3 to comprise an insert of about 11 kb in size.
  • Figure 4 is an illustrative Western blot analysis of proteins from E. coli clone LE392/8B6, which expresses the 30 kD OMP antigen.
  • Figure 5 shows a similar Western blot analysis of a phage lysate from a clone expressing the HMWP OMP, designated LE 392/17C7, probed with monoclonal antibody 17C7.
  • Lanes C-E comprise phage lysate proteins from clone LE392/17C7. These lanes exhibit slight reactivity in the very high molecular range.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Immunology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Oncology (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Veterinary Medicine (AREA)
  • Communicable Diseases (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

La présente invention se rapporte à des protéines antigéniques sélectionnées obtenues par les membranes externes de Moraxella catarrhalis, dont on a découvert qu'elles présentent une variété de propriétés utiles. Ces protéines, appelées "protéines de membrane externe", se caractérisent en ce qu'elles possèdent des masses moléculaires d'environ 30 kD, 80kD et entre environ 200 et 700 kD, respectivement. Des études ont démontré que des anticorps monoclonaux dirigés contre ces protéines confèrent un effet protecteur contre l'infection par des organismes Moraxella catarrhalis dans les modèles animaux, démontrant l'utilité potentielle de tels anticorps à conférer une immunité passive ainsi que l'utilité éventuelle de ces "protéines de membrane externe", ou variantes de celles-ci, dans la préparation de vaccins. L'invention se rapporte également à des segments d'ADN codant ces "protéines de membrane externe" à des procédés de préparation des antigènes, ou leurs variantes, par l'application des antigènes, ou leurs variantes, par l'application de techniques de génie génétique, ainsi que des procédés et réalisations de diagnostics.
EP92918273A 1991-08-15 1992-08-14 Procedes et compositions relatifs a des antigenes utiles de moraxella catarrhalis Expired - Lifetime EP0612250B1 (fr)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US745591 1991-08-15
US07/745,591 US5552146A (en) 1991-08-15 1991-08-15 Methods and compositions relating to useful antigens of Moraxella catarrhalis
PCT/US1992/006869 WO1993003761A1 (fr) 1991-08-15 1992-08-14 PROCEDES ET COMPOSITIONS RELATIFS A DES ANTIGENES UTILES DE $i(MORAXELLA CATARRHALIS)

Publications (2)

Publication Number Publication Date
EP0612250A1 true EP0612250A1 (fr) 1994-08-31
EP0612250B1 EP0612250B1 (fr) 1996-07-24

Family

ID=24997358

Family Applications (1)

Application Number Title Priority Date Filing Date
EP92918273A Expired - Lifetime EP0612250B1 (fr) 1991-08-15 1992-08-14 Procedes et compositions relatifs a des antigenes utiles de moraxella catarrhalis

Country Status (14)

Country Link
US (3) US5552146A (fr)
EP (1) EP0612250B1 (fr)
JP (1) JP3499868B2 (fr)
KR (1) KR100271888B1 (fr)
AT (1) ATE140627T1 (fr)
AU (1) AU666329B2 (fr)
CA (1) CA2115565C (fr)
DE (1) DE69212495T2 (fr)
DK (1) DK0612250T3 (fr)
ES (1) ES2092696T3 (fr)
FI (1) FI106844B (fr)
GR (1) GR3021423T3 (fr)
NO (2) NO308413B1 (fr)
WO (1) WO1993003761A1 (fr)

Families Citing this family (46)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2693654B1 (fr) * 1992-07-20 1994-08-26 Oreal Médicament, notamment immunomodulateur, contenant des enveloppes ou fractions d'enveloppes de bactéries filamenteuses non photosynthétiques et non fructifiantes, et sa préparation.
ES2145072T3 (es) 1993-05-13 2000-07-01 American Cyanamid Co Preparacion y usos de proteinas de membranas externas carentes de los de cocos gramnegativos.
US5712118A (en) * 1993-09-29 1998-01-27 Research Foundation Of State University Of New York Vaccine for branhamella catarrhalis
US5607846A (en) * 1994-05-17 1997-03-04 Research Foundation Of State University Of New York Vaccine for moraxella catarrhalis
US6121427A (en) * 1994-10-24 2000-09-19 Connaught Laboratories Limited Major outer membrane protein CD of branhamella
US6440425B1 (en) * 1995-05-01 2002-08-27 Aventis Pasteur Limited High molecular weight major outer membrane protein of moraxella
US6335018B1 (en) 1995-05-01 2002-01-01 Aventis Pasteur Limited High molecular weight major outer membrane protein of moraxella
US6290970B1 (en) * 1995-10-11 2001-09-18 Aventis Pasteur Limited Transferrin receptor protein of Moraxella
US7341727B1 (en) * 1996-05-03 2008-03-11 Emergent Product Development Gaithersburg Inc. M. catarrhalis outer membrane protein-106 polypeptide, methods of eliciting an immune response comprising same
KR100500000B1 (ko) * 1996-05-03 2005-12-12 안텍스 바이오로직스 인코퍼레이션 모락셀라카타르할리스외측막단백질-16폴리펩티드,이의유전자서열및이의용도
US6004562A (en) * 1996-08-16 1999-12-21 The Research Foundation Of The State University Of New York Outer membrane protein B1 of Moraxella catarrhalis
US6558677B2 (en) * 1996-10-15 2003-05-06 Wendell D. Zollinger Vaccine against gram negative bacteria
BR9714160A (pt) 1996-12-20 2000-05-02 Univ Texas Antìgenos uspa1 e uspa2 de moraxella catarrhalis
GB9810084D0 (en) 1998-05-11 1998-07-08 Cortecs Uk Ltd Proteins
US6627728B1 (en) 1998-05-12 2003-09-30 Smithkline Beecham Biologicals S.A. Compounds from moraxella catarrhalis
GB9810285D0 (en) * 1998-05-13 1998-07-15 Smithkline Beecham Biolog Novel compounds
GB9812440D0 (en) * 1998-06-09 1998-08-05 Smithkline Beecham Biolog Novel compounds
MY125202A (en) 1999-03-19 2006-07-31 Smithkline Beecham Biologicals S A Vaccine
GB9914945D0 (en) * 1999-06-25 1999-08-25 Smithkline Beecham Biolog Novel compounds
GB9921691D0 (en) * 1999-09-14 1999-11-17 Smithkline Beecham Sa Novel compounds
GB0022742D0 (en) 2000-09-15 2000-11-01 Smithkline Beecham Biolog Vaccine
GB0103171D0 (en) 2001-02-08 2001-03-28 Smithkline Beecham Biolog Vaccine composition
KR101239242B1 (ko) 2002-08-02 2013-03-11 글락소스미스클라인 바이오로지칼즈 에스.에이. 항원 조합물을 포함하는 나이세리아 백신 조성물
GB0220194D0 (en) 2002-08-30 2002-10-09 Chiron Spa Improved vesicles
GB0424092D0 (en) 2004-10-29 2004-12-01 Chiron Srl Immunogenic bacterial vesicles with outer membrane proteins
IL308456A (en) 2005-04-08 2024-01-01 Wyeth Llc A multivalent pneumomuroral protein-polysaccharide conjugate preparation
US7709001B2 (en) 2005-04-08 2010-05-04 Wyeth Llc Multivalent pneumococcal polysaccharide-protein conjugate composition
TWI457133B (zh) 2005-12-13 2014-10-21 Glaxosmithkline Biolog Sa 新穎組合物
PT1962899E (pt) 2005-12-22 2011-10-19 Glaxosmithkline Biolog Sa Vacina conjugada polissacarídica pneumocócica
GB0607088D0 (en) 2006-04-07 2006-05-17 Glaxosmithkline Biolog Sa Vaccine
CA2636566C (fr) 2006-01-17 2018-03-13 Arne Forsgren Nouvelle proteine de haemophilus influenzae a surface exposee (proteine; pe)
BRPI0813644B8 (pt) 2007-06-26 2021-05-25 Glaxosmithkline Biologicals Sa composição imunogênica, vacina, processo para fabricar a mesma, e, uso da composição imunogênica ou vacina
CA2726465A1 (fr) 2008-05-30 2009-12-30 Wendell David Zollinger Vaccin comprenant des vesicules de membrane externe native polyvalent contre des maladies a meningocoques, procede de fabrication et d'utilisation de celui-ci
GB201003922D0 (en) 2010-03-09 2010-04-21 Glaxosmithkline Biolog Sa Conjugation process
GB201003924D0 (en) 2010-03-09 2010-04-21 Glaxosmithkline Biolog Sa Immunogenic composition
CA2792683A1 (fr) 2010-03-10 2011-09-15 Glaxosmithkline Biologicals S.A. Composition de vaccin fhbp neisserial
GB201103836D0 (en) 2011-03-07 2011-04-20 Glaxosmithkline Biolog Sa Conjugation process
GB201518684D0 (en) 2015-10-21 2015-12-02 Glaxosmithkline Biolog Sa Vaccine
GB201621686D0 (en) 2016-12-20 2017-02-01 Glaxosmithkline Biologicals Sa Novel methods for inducing an immune response
BR112019025193A2 (pt) 2017-05-30 2020-06-23 Glaxosmithkline Biologicals S.A. Métodos de fabricação de um adjuvante lipossomal, de fabricação de um concentrado lipossomal, para a preparação de uma composição imunogênica com adjuvante e para a preparação de uma solução, adjuvante lipossomal, composição imunogênica com adjuvante, e, solução
JP2021504424A (ja) 2017-12-01 2021-02-15 グラクソスミスクライン バイオロジカルズ ソシエテ アノニム サポニン精製
US20210283238A1 (en) 2018-08-07 2021-09-16 Glaxosmithkline Biologicals Sa Novel processes and vaccines
EP3886901A1 (fr) 2018-11-29 2021-10-06 GlaxoSmithKline Biologicals S.A. Procédés de fabrication d'un adjuvant
US20220235095A1 (en) 2019-06-05 2022-07-28 Glaxosmithkline Biologicals Sa Saponin purification
GB202205833D0 (en) 2022-04-21 2022-06-08 Glaxosmithkline Biologicals Sa Bacteriophage
WO2024017827A1 (fr) 2022-07-19 2024-01-25 Glaxosmithkline Biologicals Sa Procédé continu de production de vaccin

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5030556A (en) * 1989-03-30 1991-07-09 Danielle Beaulieu Species-specific DNNA probe for the detection of Branhamella catarrhalis

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO9303761A1 *

Also Published As

Publication number Publication date
AU2487892A (en) 1993-03-16
DE69212495T2 (de) 1997-03-06
FI106844B (fi) 2001-04-30
WO1993003761A1 (fr) 1993-03-04
DE69212495D1 (de) 1996-08-29
EP0612250B1 (fr) 1996-07-24
NO940502L (no) 1994-03-28
JPH07501210A (ja) 1995-02-09
NO308413B1 (no) 2000-09-11
NO20002413D0 (no) 2000-05-09
FI940681A0 (fi) 1994-02-14
US5759813A (en) 1998-06-02
CA2115565C (fr) 2007-08-07
US5599693A (en) 1997-02-04
KR100271888B1 (ko) 2000-11-15
FI940681A (fi) 1994-04-07
GR3021423T3 (en) 1997-01-31
NO940502D0 (no) 1994-02-14
AU666329B2 (en) 1996-02-08
DK0612250T3 (da) 1996-11-25
ES2092696T3 (es) 1996-12-01
JP3499868B2 (ja) 2004-02-23
US5552146A (en) 1996-09-03
NO20002413L (no) 2000-05-09
CA2115565A1 (fr) 1993-03-04
ATE140627T1 (de) 1996-08-15

Similar Documents

Publication Publication Date Title
US5759813A (en) Methods and compositions relating to useful antigens of moraxella catarrhalis
US6420134B1 (en) Vaccines for nontypable haemophilus influenzae
JP2907552B2 (ja) ヘモフィルス外膜タンパク質
JP2009142276A (ja) モラクセラ・カタラーリスのuspa1及びuspa2抗原
US5993826A (en) Methods and compositions relating to useful antigens of moraxella catarrhalis
JP2002525110A (ja) モラクセラ・カタラーリスのタンパク質、核酸配列およびその使用
AU740481B2 (en) Defining epitopes of the outer membrane protein CopB of moraxella catarrhalis
US5380655A (en) Methods and compositions for the production of haemophilus influenzae type B major outer membrane protein antigens
AU715860B2 (en) H. Influenzae HxuB and HxuC genes, proteins and methods of use
JPH0213376A (ja) 緑膿菌に対するモノクローナル抗体、ならびにその調製および使用
US20020164354A1 (en) High molecular weight surface proteins of non-typeable haemphilus
US20030007981A1 (en) Cloning and expression of Haemophilus somnus transferrin-binding proteins
US7101989B1 (en) DsrA protein and polynucleotides encoding the same
US6887482B2 (en) Antigenic iron repressible proteins from N. meningitidis related to the hemolysin family of toxins
CHONG et al. Sommaire du brevet 2149319
CHONG et al. Patent 2149319 Summary
MXPA97009557A (en) Members of streptococal thermal shock proteins of the hs family

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 19940315

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LI LU NL SE

17Q First examination report despatched

Effective date: 19950512

GRAH Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOS IGRA

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LI LU NL SE

REF Corresponds to:

Ref document number: 140627

Country of ref document: AT

Date of ref document: 19960815

Kind code of ref document: T

GRAH Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOS IGRA

REF Corresponds to:

Ref document number: 69212495

Country of ref document: DE

Date of ref document: 19960829

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

Free format text: 69232

ET Fr: translation filed
ITF It: translation for a ep patent filed

Owner name: JACOBACCI & PERANI S.P.A.

REG Reference to a national code

Ref country code: CH

Ref legal event code: NV

Representative=s name: HEPP, WENGER & RYFFEL AG

REG Reference to a national code

Ref country code: DK

Ref legal event code: T3

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2092696

Country of ref document: ES

Kind code of ref document: T3

REG Reference to a national code

Ref country code: GR

Ref legal event code: FG4A

Free format text: 3021423

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

26N No opposition filed
REG Reference to a national code

Ref country code: GB

Ref legal event code: IF02

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: DE

Payment date: 20070809

Year of fee payment: 16

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: IE

Payment date: 20070813

Year of fee payment: 16

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: DK

Payment date: 20070815

Year of fee payment: 16

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: LU

Payment date: 20070816

Year of fee payment: 16

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: ES

Payment date: 20070926

Year of fee payment: 16

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: CH

Payment date: 20070815

Year of fee payment: 16

Ref country code: AT

Payment date: 20070813

Year of fee payment: 16

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GB

Payment date: 20070809

Year of fee payment: 16

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: SE

Payment date: 20070807

Year of fee payment: 16

Ref country code: NL

Payment date: 20070805

Year of fee payment: 16

Ref country code: IT

Payment date: 20070828

Year of fee payment: 16

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: BE

Payment date: 20071018

Year of fee payment: 16

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: FR

Payment date: 20070808

Year of fee payment: 16

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GR

Payment date: 20070813

Year of fee payment: 16

REG Reference to a national code

Ref country code: DK

Ref legal event code: EBP

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

EUG Se: european patent has lapsed
GBPC Gb: european patent ceased through non-payment of renewal fee

Effective date: 20080814

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: AT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20080814

NLV4 Nl: lapsed or anulled due to non-payment of the annual fee

Effective date: 20090301

REG Reference to a national code

Ref country code: IE

Ref legal event code: MM4A

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: NL

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20090301

REG Reference to a national code

Ref country code: FR

Ref legal event code: ST

Effective date: 20090430

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20080831

Ref country code: GR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20090304

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20080831

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20080814

Ref country code: DK

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20080831

Ref country code: BE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20080831

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20080814

Ref country code: FR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20080901

Ref country code: DE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20090303

REG Reference to a national code

Ref country code: ES

Ref legal event code: FD2A

Effective date: 20080816

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: GB

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20080814

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: ES

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20080816

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20080814

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20080815